Journal Pre-proof Metabolomic-transcriptomic landscape of 8-epidiosbulbin E acetate -a major diterpenoid lactone from Dioscorea bulbifera tuber induces hepatotoxicity Wei Shi, Yan Jiang, Dong-Sheng Zhao, Li-Long Jiang, Feng-Jie Liu, Zi-Tian Wu, Zhuo-Qing Li, Ling-Li Wang, Jing Zhou, Ping Li, Hui-Jun Li PII:
S0278-6915(19)30677-5
DOI:
https://doi.org/10.1016/j.fct.2019.110887
Reference:
FCT 110887
To appear in:
Food and Chemical Toxicology
Received Date: 9 April 2019 Revised Date:
8 October 2019
Accepted Date: 12 October 2019
Please cite this article as: Shi, W., Jiang, Y., Zhao, D.-S., Jiang, L.-L., Liu, F.-J., Wu, Z.-T., Li, Z.-Q., Wang, L.-L., Zhou, J., Li, P., Li, H.-J., Metabolomic-transcriptomic landscape of 8-epidiosbulbin E acetate -a major diterpenoid lactone from Dioscorea bulbifera tuber induces hepatotoxicity, Food and Chemical Toxicology (2019), doi: https://doi.org/10.1016/j.fct.2019.110887. This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. Please note that, during the production process, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain. © 2019 Published by Elsevier Ltd.
1
Metabolomic-transcriptomic landscape of 8-epidiosbulbin E acetate
2
-a major diterpenoid lactone from Dioscorea bulbifera tuber induces
3
hepatotoxicity
4 a
b,*
a
a
a
5
Wei Shi , Yan Jiang , Dong-Sheng Zhao , Li-Long Jiang , Feng-Jie Liu , Zi-Tian
6
Wu , Zhuo-Qing Li , Ling-Li Wang , Jing Zhou , Ping Li , Hui-Jun Li
a
a
a
a
a
a,*
7 8 9 10
a
State Key Laboratory of Natural Medicines, China Pharmaceutical University,
Nanjing, 210009, China b
Nanjing Forestry University, Nanjing, 210037, China
11 12
*Corresponding Authors:
13
Hui-Jun Li, PhD
14
State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24
15
Tongjia Lane, Nanjing 210009, China.
16
E-mail:
[email protected].
17
Tel.: +86 25 83271382; Fax: +86 25 83271379.
18
Yan Jiang, PhD
19
Nanjing Forestry University, No. 159 Longpan Road, Nanjing 210037, China.
20
E-mail:
[email protected].
21
Tel.: +86 25 83271382; Fax: +86 25 83271379.
22 1
23
ABSTRACT
24
Studies have shown that 8-epidiosbulbin E acetate (EEA), a major diterpenoid lactone
25
in the tuber of Dioscorea bulbifera, can induce hepatotoxicity in vivo. However, the
26
underlying mechanisms remain unknown. Using the integrated transcriptomic and
27
metabolomics method, in this study we investigated the global effect of EEA exposure
28
on the transcriptomic and metabolomic profiles in mice. The abundance of 7131
29
genes and 42 metabolites in the liver, as well as 43 metabolites in the serum were
30
altered. It should be noted that EEA mainly damaged hepatic cells through the
31
aberrant regulation of multiple systems primarily including bile acid metabolism, and
32
taurine and hypotaurine metabolism. In addition, an imbalance of bile acid
33
metabolism was found to play a key pat in response to EEA-triggered hepatotoxicity.
34
In summary, these findings contributed to understanding the underlying mechanisms
35
of EEA hepatotoxicity.
36 37
Keywords: 8-Epidiosbulbin E acetate; Transcriptomics; Metabolomics; Hepatoto
38
xicity; Bile acid metabolism
2
39
1. Introduction
40
Dioscorea bulbifera tuber (DBT), a medicinal herb belonging to the
41
Dioscoreaceae family, has been widely used in China and exhibits a variety of
42
pharmacological activities, including goiter inhibitory, anti-inflammatory, and
43
antitumor effects (Tang, 1995; Rao et al., 2010; Liu et al., 2011; Wang et al., 2012).
44
Despite a wide array of therapeutic values, the hepatotoxicity of DBT has been a
45
critical safety issue restricting its application (Liu, 2002; Huang et al., 2013). Liver
46
injury cases have frequently been found to be associated with the consumption of
47
DBT in clinical practice (Yang et al., 2006; Lu and Wu, 2014). Studies have attributed
48
the DBT-induced hepatotoxicity to different causes, including oxidative damage to
49
hepatic mitochondria or bile acid (BA) metabolic disorders (Wang et al., 2010; Wang
50
et al., 2011; Zhao et al., 2017; Xiong et al., 2017).
51
A number of diterpenoid lactones (DLs) have been isolated and identified from
52
DBT; the major DLs found in DBT were reportedly 8-epidiosbulbin E acetate (EEA)
53
and diosbulbin B (DIOB) (Kawasaki et al., 1968; Murray et al., 1984; Gao et al.,
54
2002). Our preliminary study revealed that DIOB and EEA played an important role
55
in the hepatotoxicity of DBT (Shi et al., 2018). However, compared with abundant
56
investigations on DIOB, few studies have been associated with EEA. Lin et al.
57
demonstrated that administration of EEA could cause severe acute hepatotoxicity in
58
vivo and the observed liver injury required cytochromes P450-mediated metabolism
59
(Lin et al., 2015). The electrophilic intermediate generated by metabolic activation of
60
the furan ring is responsible for EEA-induced hepatotoxicity (Lin et al., 2016). 3
61
Furthermore, the cysteine- and lysine-based protein adduction with the reactive
62
intermediate was found both in vitro and in vivo, where the changes in levels of the
63
protein adductions might be related to alternations in degrees of EEA-induced
64
hepatotoxicity (Lin et al., 2016). Although these investigations facilitate a better
65
understanding of the mechanism of hepatoxic action of EEA, a consensus is still far
66
from being realized. Therefore, it is urgently necessary to reveal the underlying
67
mechanism of EEA-induced hepatic damage.
68
Generating high-throughput data from multiple technique platforms, systems
69
biology is being increasingly applied for the search of novel biomarkers and a deeper
70
understanding of the toxic effects of xenobiotics (Hua et al., 2015; Basu et al., 2017).
71
Transcriptomics, which can identify sensitive and detailed insights into the potential
72
mechanisms of toxic action at an earlier molecular level, provides a valuable platform
73
in predicting possible toxicity and further revealing the potential biomarkers in
74
toxicity (Cui et al., 2010; Li et al., 2014). Focusing on studying the multi-parametric
75
metabolic responses in living systems after pathophysiological stimuli or genetic
76
modification, metabonomics can also relate these responses to the gene expression
77
profiles and pathology results (Chen et al., 2018; Fan et al., 2016). Moreover, the
78
integration of transcriptomics and metabolomics profiles may offer a greater
79
reliability in expounding metabolic alterations and allow further elucidation toward
80
the toxic effects and mechanism (Lu et al., 2013). To the best of our knowledge, the
81
integrated application of these two platforms to characterize EEA-induced hepatic
82
toxicity has not yet been reported. 4
83
EEA has a potential hepatotoxic effect on the physiological and biological
84
functions of organisms via regulating multiple metabolic pathways to an abnormal
85
state. In the present study, an integrated analysis of general toxicity studies,
86
transcriptomics, and metabonomics approaches was applied to investigate
87
EEA-induced hepatotoxicity in mice (Fig. 1). Moreover, the pathways related to the
88
toxic effects of EEA were summarized by correlation network analysis and validated
89
by quantitative real-time PCR (qRT-PCR) and Western blot analyses. The objectives
90
of the present study were to provide a comprehensive insight into the mechanism and
91
potential biomarkers of EEA-induced hepatotoxicity.
92
93
2. Materials and methods
94
2.1. Reagents
95
The DBT was obtained from Yunnan province, China. The sample was
96
authenticated by Prof. Hui-Jun Li and deposited at State Key Laboratory of Natural
97
Medicines (China Pharmaceutical University, Nanjing, China).
98
Chromatographic grade methanol was obtained from CNW Technologies Gmbh
99
(Dussel-dorf, Germany). Chloroform and pyridine were provided by Adamas Reagent
100
Co., Ltd. (Basel, Switzerland). L-2-Chlorophenylalanine (internal standard, IS) was
101
purchased from Shanghai Heng Bo Biological Technology Co., Ltd. (Shanghai,
102
China).
103
trimethylchlorosilane (TMCS) was purchased from REGIS Technologies (Chicago,
N,O-bis
(trimethylsilyl)
trifluoroacetamide
5
(BSTFA)
with
1%
104
USA). Deionized water (18 MΩ cm) was prepared by distilled water through a
105
Milli-Q system (Massachusetts, USA). Bile acid standards were purchased from
106
Sigma-Aldrich Co. (St. Louis, MO, USA), including cholic acid (CA), lithocholic
107
acid (LCA), deoxycholic acid (DCA), ursodeoxycholic acid (UDCA), hyodeoxycholic
108
acid
109
glycochenodeoxycholic acid (GCDCA), tauroursodeoxycholic acid (TUDCA),
110
taurohyodeoxycholic acid (THDCA), taurocholic acid (TCA), taurolithocholic acid
111
(TLCA), dehydrocholic acid (DHCA, IS). EEA was isolated from DBT in our
112
laboratory and the chemical structure (Fig. S1) was confirmed by extensive
113
spectroscopic analyses. The purity of EEA was > 98%, detected by ultra-high
114
performance liquid chromatography (UPLC) using peak area normalization method.
115
Other reagents and solvents were of analytical grade.
116
2.2. Animals and treatment
(HDCA),
chenodeoxycholic
acid
(CDCA),
glycocholic
acid
(GCA),
117
Male ICR mice (4–5 weeks old; weighing: 18–22 g) were purchased from
118
Sino-British SIPPR/BK Lab Animal Ltd. (Shanghai, China). Mice were fed a standard
119
laboratory diet and given free access to tap water. They were kept in a controlled
120
room temperature (22 ± 2oC), humidity (60 ± 5%), and a 12 h dark/light cycle for at
121
least 7 days before treatment. All animal studies were performed according to the
122
Provision and General Recommendation of Chinese Experimental Animals
123
Administration Legislation and were approved by Department of Science and
124
Technology of Jiangsu Province (license number: SYXK (SU) 2016-0011).
125
The mice were orally administered EEA (150 mg/kg, suspended in 0.5% 6
126
CMC-Na, n = 10) for 36 h, and 0.5% sodium carboxymethyl cellulose (CMC-Na) was
127
used as a vehicle control (n = 10). During the time they were fasted from food, but no
128
water 12 h prior to the administration of the test suspension. Blood samples and liver
129
tissues were collected at 36 h following treatment. All biological samples were
130
lyophilized and stored at −80oC before further analysis.
131
In a separate study, mice were given EEA at 30 mg/kg (i.g.), blood and liver were
132
collected 36 h after the administration, and the serum alanine aminotransferase (ALT),
133
aspartate aminotransferase (AST), total bilirubin (TBIL), direct bilirubin (DBIL) and
134
alkaline phosphatase (ALP) levels were measured. Liver samples were collected and
135
prepared for further western blots analysis.
136
2.3. Detection of AST, ALT, TBIL, DBIL and ALP levels from serum
137
Blood was collected from the eyeball and centrifuged at 1500g for 10 min at 4oC.
138
Serum AST, ALT, TBIL, DBIL and ALP activities were measured on Cobas 8000
139
modular analyzer (Basel, Switzerland).
140
2.4. Histopathologic examination
141
Liver tissues were fixed in 10% neutral buffered formalin, paraffin processed,
142
and sectioned at 4 µm. For histological evaluation, the tissue sections were stained
143
with hematoxylin and eosin (H&E) and examined for histopathological changes under
144
the Olympus DX45 microscope (Tokyo, Japan).
145
2.5. RNA isolation, cDNA library construction and illumina deep sequencing
146
Six liver tissue samples from three biological replicates of the vehicle and 7
147
EEA-treated mice groups were used for the transcriptomic analysis. Total RNA was
148
extracted using Trizol reagent (Invitrogen, USA) according to the manufacturer’s
149
protocol. The RNA integrity was assessed using a Bioanalyzer 2100 system (Agilent
150
Technologies, USA). The RNA samples for the transcriptome analysis were prepared
151
using an Illumina kit, following the manufacturer’s recommendations. The fragments
152
were purified via agarose gel electrophoresis and enriched by PCR amplification to
153
create a cDNA library. Sequencing and data processing (including the statistical
154
analysis and selection of differentially expressed genes) were all performed following
155
the methods (Katz et al., 2010).
156
2.6. Metabolic profiling analysis
157
An aliquot of 100 µL of serum or 60 mg of liver tissue sample was placed in a 2
158
mL eppendorf (EP) tube, followed by extraction with 0.48 mL of mixture of
159
chloroform and methanol (Vmethanol : Vchloroform = 3:1). Afterwards, 20 µL of
160
L-2-chlorophenylalanine (1 mg/mL stock in dH2O) were added as internal standard,
161
and samples were vortex-mixed for 30 s. A ball mill was used to homogenize the
162
samples for 4min at 45 Hz, followed by sonication for 5 min, and then centrifugation
163
for 15 min at 28000g, 4oC. Quality control (QC) samples were prepared by pooling
164
aliquots of all samples and were processed with the same procedure as that followed
165
for the experiment samples. The extracts were dried in a vacuum concentrator at 30oC
166
for approximately 1.5 h, and then 60 µL of methoxyamine hydrochloride in pyridine
167
(20 mg/mL) was added to the residue, being derivatized at 80°C for 30 min.
168
Subsequently, 80 µL of the BSTFA regent (1% TMCS, v/v) were added into the 8
169
sample aliquots, incubated for 1.5 h at 70oC, and 10 µL FAMEs (Standard mixture of
170
fatty acid methyl esters, C8-C16:1 mg/mL; C18-C24:0.5 mg/mL in chloroform) was
171
added to the samples until cooling to the room temperature. The pooled QC sample
172
was injected five times at the beginning of the run in order to ensure system
173
equilibration and then every 10 samples to further monitor the stability of the analysis.
174
2.7. GC-TOF MS analysis
175
GC-TOF MS analysis was performed using an Agilent 7890 gas chromatograph
176
system (Agilent Technologies Inc., Santa Clara, California, USA) coupled with a
177
Pegasus HT time-of-flight mass spectrometer (LECO Corp., St. Joseph, MI, USA)
178
that included an Rxi-5Sil MS column (Restek, Bellefonte, USA), which was 30 m in
179
length and 0.25 mm in inner diameter with a film thickness of 0.25 µm. A 1 µL aliquot
180
of the analyte was injected in splitless mode. The carrier gas was helium with a 3
181
mL/min front inlet purge flow and a constant 1 mL/min gas flow rate through the
182
column. The initial temperature was maintained at 50°C for 1 min, raised to 310°C at
183
a rate of 10°C/min, and then kept for 5 min at 310°C. The injection, transfer line and
184
ion source temperatures were 280, 270, and 220°C, respectively. The energy was -70
185
eV in electron impact mode. The mass spectrometry data were acquired in full-scan
186
mode with the m/z range of 50 − 500 at a rate of 20 spectra per second after a solvent
187
delay of 370 s.
188
LECO Chroma TOF4.3X software and LECO-Fiehn Rtx5 database (Leco Corp.,
189
St. Joseph, MI, USA) were used for the raw data acquisition and processing, such as
190
the raw peak exacting, raw peak exacting, data baseline filtering and calibration, peak 9
191
alignment, deconvolution analysis, peak identification, and peak area integration. The
192
retention time index method was used for peak identification, with a tolerance value
193
of 5000.
194
Principal
component
analysis
(PCA)
and
orthogonal
partial
195
least-squares-discriminate analysis (OPLS-DA) were performed using SIMCA
196
version
197
(http://www.metaboanalyst.ca/). Statistical analyses were conducted by SPSS
198
software version 19.0 (IBM Corp., Armonk, USA). On the basis of variable
199
importance in the projection (VIP) scores > 1.0 obtained from the OPLS-DA model
200
and p values < 0.05 evaluated by the student’s t-test, a set of discriminating
201
metabolites were determined (Afanador et al., 2013; Matthews et al., 1985). Heat
202
maps and hierarchical cluster analyses were conducted using MeV 4.6.0 software. The
203
pathway mapping of the serum was analyzed with MetScape 3 (Karnovsky et al.,
204
2011).
205
2.8. Integrated pathway analysis
14.0.1
(Umetrics,
Sweden)
and
MetaboAnalyst
4.0
206
To identify the related pathways among the metabolites and genes,
207
MetaboAnalyst 4.0 (http://www.metaboanalyst.ca/) was utilized to further holistic
208
pathway analysis. Hypergeometric tests were calculated to evaluate the enrichment
209
analysis between metabolome and transcriptome data, and the topology analysis
210
(Degree Centrality) were applied to evaluate whether a given gene or metabolite plays
211
an important role in a biological response based on its position in the pathway (Chong
212
et al., 2018). 10
213
2.9. Real-time RT-PCR
214
To examine the accuracy and reproducibility of the Illumina RNA-Seq results,
215
qRT-PCR assays were performed with gene-specific primers (Suo et al., 2018). Total
216
RNA was extracted from EEA-treated mice using the TRIzol reagent (Invirtrogen,
217
USA) and qRT-PCR was performed using SYBR Premix Ex Taq II (TaKaRa, Japan)
218
following the manufacturer’s protocol. qRT-PCR was conducted on Applied
219
Biosystems 7,300 machine (Carlsbad, USA) as follows: 3 min at 95°C for
220
denaturation, followed by 40 cycles of 7 s at 95°C for denaturation, 10 s at 57°C for
221
annealing, and 30 s at 72°C for extension. For each target gene, triplicate reactions
222
were performed. Relative gene expression levels were calculated from cycle threshold
223
values using the 2−∆∆Ct method (Livak and Schmittgen, 2001). The sequences of
224
specific primers used for qRT-PCR are listed in Table 1.
225
2.10. Western blotting assay
226
Western blot analysis was performed as previously described with minor
227
modifications (Renaud et al., 2011). Liver homogenates were prepared in radio
228
immunoprecipitation assay (RIPA) buffer. Protein concentrations were determined
229
using bicinchonininc acid (BCA) assay method according to the manufacturer's
230
instructions (Beyotime, China). Samples were subjected to polyacrylamide gel
231
electrophoresis, transferred onto a polyvinylidene difluoride membrane, and probed
232
with the respective primary and secondary antibodies. Membranes were stripped and
233
reprobed with a β-actin antibody as the loading control. Proteins were detected using
11
234
chemiluminescence.
235
2.11. UPLC-QqQ-MS profiling and MS conditions for BAs analysis
236
Metabolomic profiling analysis of BAs was performed by following our
237
previously published method (Zhao et al., 2017). Liver samples (60 mg) were mixed
238
with 400 µL of methanol containing 10 µL IS. The mixture was vortexed for 5 min,
239
followed by centrifugation at 28000g for 10 min at 4°C, and the supernatant was
240
analyzed by UPLC-QqQ/MS.
241
3. Results
242
3.1. Hepatotoxicity of EEA in mice
243
Histopathologic analysis (n = 5 per group) showed inflammatory cell infiltration,
244
hepatic cell necrosis and focal necrosis (Fig. 2A, B) in mice liver i.g. administered
245
EEA at 150 mg/kg. Additionally, the serum ALT/AST/TBIL/DBIL levels significantly
246
increased (p <0.01), compared with 30.1 ± 4.4 U/L (ALT), 105.0 ± 27.3 U/L (AST),
247
1.1 ± 0.2 U/L (TBIL) and 0.9 ± 0.2 U/L (DBIL) in mice treated with vehicle (Fig.
248
2C-F). Additionally, the serum ALP levels were also significantly increased (p <0.05)
249
(Fig. 2G). Compared with mice treated with EEA (150 mg/kg), no changes were
250
observed in the levels of ALT/AST/TBIL/DBIL/ALP in mice administered EEA (30
251
mg/kg) (Fig. 2C-G). These findings indicated that EEA administered to the mice at
252
the dose of 150 mg/kg caused potential hepatotoxicity.
253
3.2. Differentially expressed genes (DEGs) identification and selection
12
254
In order to identify hepatotoxicity-related candidate genes that responded to EEA
255
(150 mg/kg) infection, four transcriptome profiles were performed. First, the
256
expression level of each gene was normalized using the fragments per kilobase of
257
transcript per million mapped reads (FPKM) (Trapnell et al., 2010). The genes with
258
data false discovery rate (FDR) < 0.001 and estimated absolute log2fold change
259
(log2FC) ≥ 1 in sequence counts across libraries were then considered as significant
260
DEGs. Finally, a total of 7131 DEGs were identified between EEA-treated groups and
261
the control groups. Of these genes, 3749 genes were up-regulated and 3382 genes
262
were down-regulated in the liver of mice given EEA at 150 mg/kg.
263
3.3. Gene annotation and the functional classification of DEGs
264
To gain additional insights into the function of DEGs, GO and KEGG databases
265
were used to annotate and classify DEG functions. DEGs overrepresented in the two
266
groups using GO analysis (Fig. S3) suggested that the cofactor metabolic process was
267
the most significantly enriched biological function.
268
The KEGG database was used to further understand the biological functions and
269
pathways of DEGs. Based on the KEGG pathways enrichment analysis, metabolic
270
pathways were found to be one of the most over-represented processes in mice
271
exposed to EEA treatment (150 mg/kg), suggesting that enzymatic reaction and
272
enzymes might play key roles in the development of EEA-induced liver injury. The
273
list of over-represented KEGG pathways in mice exposed to EEA (150 mg/kg) is
274
presented in Table S1.
13
275
3.4. Metabolic profiling analysis of serum and livers
276
The stability and repeatability of the GC-TOF MS system for large-scale sample
277
analysis were confirmed by the analysis of pooled QC samples and the retention time
278
of the internal standard. The serum and liver metabolic profiling analysis was
279
performed according to the proposed approach (Dunn et al., 2011). Typical total ion
280
chromatograms (TICs) of serum and livers samples from the control and EEA (150
281
mg/kg) groups are presented in Fig. S4 (A: serum; B: livers). A total of 562 and 722
282
metabolite peaks in serum and livers were respectively extracted through GC-TOF
283
MS (Fig. S5). To obtain a comprehensive view of the metabonome, PCA was
284
performed to visualize the trends and outliers in the data for the control and EEA (150
285
mg/kg) groups. The RSDs of the retention time of L-2-chlorophenylalanine (IS) in
286
serum and livers were 0.0048% and 0.0132%, respectively. The PCA also showed a
287
tight grouping of the QC samples (Fig. 3A and Fig. 4A). These data demonstrate a
288
high reproducibility of the method. The score plots revealed that there were no
289
outliers and that the two groups were clearly separated (Fig. 3 and Fig. 4).
290
To study the changes in metabolites in mice after EEA (150 mg/kg)
291
administration, the liver and serum samples were analyzed by OPLS-DA to identify
292
the variables responsible for the differences among groups (Bylesjö et al., 2010; Chan
293
et al., 2009). In the OPLS-DA score plots (Fig. 3B and Fig. 4B), the EEA groups
294
exhibited a tendency to deviate from the control groups, revealing a visible
295
perturbation of the metabolic profiles in EEA-150 mg/kg treated groups. The models
296
were validated by the permutation test. The R2 values of serum and liver samples in 14
297
the OPLS-DA models were 0.879 and 0.755 (Fig. 3C and Fig. 4C), respectively,
298
indicating an excellent prediction.
299
To identify the variables responsible for this large separation, the importance of
300
the VIP statistics from OPLS-DA modelling and t-tests (P< 0.05) between the two
301
groups was used to pre-select variables. The GC-TOF MS spectra of the metabolites
302
were analyzed based on mass spectra libraries, and the metabolites had to meet the
303
conditions VIP > 1 and P < 0.05. In the EEA-treated groups (150 mg/kg), a total of 43
304
and 42 metabolites were, respectively, identified in the serum and liver samples (Table
305
S2). Additionally, heat maps were created based on the average fold change of each
306
metabolite to intuitively evaluate the variation tendency of the metabolite
307
concentrations between the control and EEA (150 mg/kg)-treated groups. As shown in
308
Fig.5, 6 metabolites had decreased in the serum and 7 metabolites had increased in the
309
livers. Compared with the control group, one metabolite (3-hydroxybutyric acid) was
310
decreased, while six metabolites (2-hydroxybutanoic acid, uracil, N-methyl-dl-alanine,
311
fumaric acid, L-malic acid and citric acid) had an opposite tendency in both serum
312
and livers. Subsequently, the metabolic pathways analysis was performed based on
313
the metabolites in serum and livers. As a web-based server supporting pathway
314
analysis, MetaboAnalyst 4.0 (www.metaboanalyst.ca) integrates enrichment analysis
315
and pathway topology analysis to discover the significant and relevant pathways
316
affected by EEA administration. Three metabolic pathways (valine, leucine and
317
isoleucine biosynthesis phenylalanine, tyrosine and tryptophan biosynthesis and
318
D-Glutamine and D-glutamate metabolism) participated in the most relevant 15
319
pathways affected by EEA, with an impact value of 1.0 in serum (Fig. 6A). Figure 6B
320
demonstrates that elevated phenylalanine, tyrosine and tryptophan biosynthesis was
321
filtered out as significant metabolic pathways marking the impact of EEA in livers.
322
Moreover, compared with the control group, several other important pathways,
323
including citrate cycle (TCA cycle), phenylalanine metabolism, β-alanine metabolism
324
and taurine and hypotaurine metabolism, were activated in both serum and livers. In
325
the present study, our results illustrated that the pathways of EEA-induced
326
hepatotoxicity were related to purine metabolism, glutathione metabolism, as well as
327
primary bile acid biosynthesis in serum or livers of mice treated with EEA at 150
328
mg/kg.
329
3.5. Key pathways related to EEA-induced hepatotoxicity
330
MetaboAnalyst 4.0 was used to visualize and interpret the metabolomic and gene
331
expression profiling data in livers (Chong et al., 2010), allowing us to build and
332
analyze networks of genes and compounds, identify enriched pathways from the
333
differential expression profiling data, and visualize changes in metabolite data.
334
Results indicate that 79 signaling pathways played important roles in EEA-induced
335
liver injury, and the most important 20 pathways in mice exposed to EEA (150 mg/kg)
336
are shown in Fig.7. Amino acid metabolism (valine, leucine and isoleucine
337
degradation, arginine and proline metabolism, tryptophan metabolism, D-glutamine
338
and D-glutamate metabolism, glutathione metabolism and taurine and hypotaurine
339
metabolism), fatty acid metabolism (fatty acid metabolism, primary bile acid
340
biosynthesis and arachidonic acid metabolism), carbohydrate metabolism (TCA cycle, 16
341
propanoate metabolism, pentose and glucuronate interconversions, ascorbate and
342
aldarate metabolism, galactose metabolism and glyoxylate and dicarboxylate
343
metabolism)
344
metabolism-cytochrome P450, metabolism of xenobiotics by cytochrome P450 and
345
drug metabolism-other enzymes) were found to play important roles in EEA-induced
346
hepatotoxicity. Other pathways were involved in metabolism of cofactors, vitamins
347
and nucleotide metabolism, including “retinol metabolism,” “purine metabolism,” and
348
“pyrimidine metabolism.”
349
3.6. Validation of RNA-Seq data by qRT-PCR
and
xenobiotics
biodegradation
and
metabolism
(drug
350
To validate the RNA-Seq data, 10 differentially expressed genes were chosen for
351
a gene expression analysis via qRT-PCR (Fig.S6). In total, these 10 genes exhibited
352
consistent expression patterns between the RNA-Seq data and qRT-PCR. As
353
mentioned above, both data sets strongly correlated in the present study.
354
3.7. Regulation of FN1, UGT1a1, CYP1B1, CYP1A1, CYP2E1, CYP8B1, CYP7A1,
355
BAAT, NTCP, BSEP and MRP3 protein expression in the livers of mice treated with
356
EEA
357
The integrated analysis of the results from transcriptome and metabolome
358
profiles indicated that down-regulation of key enzymes and disorder of BA
359
metabolism in the liver played a pivotal role in the hepatotoxicity of EEA. The results
360
showed downregulation of CYP7A1 and CYP8B1 protein expression in livers from
361
EEA(150 mg/kg)-treated mice compared to vehicle groups. In addition, three 17
362
important BA transporters - NTCP, MRP3, and BSEP protein expression - were also
363
measured with a significant down-regulation of NTCP, MRP3, and BSEP protein
364
expression observed between the control and treatment groups (150 mg/kg).
365
Additionally, CYP1A1 was found to be significantly up-regulated in EEA-treated
366
mice livers. Western blot results are shown in Fig.8A.
367
As shown in Fig. 8B, no significant changes were observed in the levels of
368
CYP7A1, CYP1A1, NTCP and BSEP protein expression in EEA (30 mg/kg) groups,
369
which were in accordance with the results of biochemical indices serum
370
determination.
371
3.8. Quantitative analysis of BAs
372
By combining metabolomics and transcriptomics data, studying the alternation in
373
the BA metabolism pathway after EEA (150 mg/kg) treatment has become an
374
interesting topic. Therefore, concentrations of 12 individual bile acids - including 6
375
free bile acids, 2 glycine-conjugated bile acids, and 4 taurine-conjugated bile acids -
376
were simultaneously quantified in the liver of the control and EEA treated groups
377
(150 mg/kg) using our published method (Zhao et al., 2017). As shown in Fig. 9,
378
compared with the control group, the concentrations of 2 bile acids were significantly
379
increased, and one (CDCA) significantly decreased in mice liver. Among these bile
380
acids, bile acids such as TCA, CA and GCA exhibited the highest increases of 6.4-,
381
3.8-, and 5.7-fold, respectively. Most importantly, the taurine conjugated bile acid
382
TCA and free bile acid CA were both significantly elevated in the EEA-treated groups
383
(p < 0.01), indicating that, along with CDCA, bile acids including TCA and CA could 18
384
be considered as biomarkers of EEA induced liver injury.
385
4. Discussion
386
The EEA-associated hepatotoxicity has been well recognized in experimental
387
animals (Lin et al., 2015). However, its underlying mechanism of toxicity remains
388
largely unexplored to date. In this work, we utilized the integrated transcriptomics and
389
metabolomics approaches to identify the alterations of genes and metabolic profiles in
390
the livers of mice with EEA-induced liver injury. Mice developed hepatotoxicity
391
following 36 h administration of 150 mg/kg of EEA, which could be ascribed to the
392
down-regulation of key enzymes and disorder of BA metabolism in the liver.
393
Serum levels of ALT, AST and bilirubin are most commonly used in current
394
clinical practice, especially for acute hepatocellular injury (Whitfield et al., 1985;
395
Van-swelm et al., 2014). The hepatotoxicity induced by a single dose of EEA at 100
396
mg/kg reached a maximum at 36 h post treatment, with inflammatory cell infiltration
397
being observed (Lin et al., 2015). Correlating well with this evidence, our data
398
showed that the levels of ALT and AST were observed with the elevation in serum
399
and confirmed by inflammatory cell infiltration and focal necrosis in the liver of mice
400
given EEA at 150 mg/kg. Remarkably, we found that TBIL and DBIL levels were also
401
sensitive biomarkers, signifying the mechanism of EEA-induced liver injury. More
402
importantly, the serum biochemical parameters showed significant elevation in the
403
levels of ALP, indicating that the administration of EEA might influence the BAs
404
metabolism of the liver (Qu et al., 2017, Fuchs et al., 2017).
19
405
The liver is a multifunctional organ that is involved in various enzymatic
406
metabolic activities. Therefore, damage to this important organ by a hepatotoxic agent
407
will lead to a disturbance in the body metabolism (Ramadori et al., 2015; McEuen et
408
al., 2017). At 36 h after EEA (150 mg/kg) treatment, the expression of 625 genes
409
involved in the metabolic pathways was found to be significantly regulated. In
410
addition, drug metabolism cytochrome P450 and retinol metabolism also showed
411
significant change. Cytochrome P450 3A4 had been demonstrated to play critical
412
roles in EEA transformation to the corresponding cis-enedial intermediate, which
413
triggered the EEA-involved hepatotoxicity (Lin et al., 2016). The integrated analysis
414
results showed that the pathway “Drug metabolism – cytochrome P450” and
415
“Metabolism of xenobiotics by cytochrome P450” (Fig. 7) were found to be the
416
significant changed pathway in the model. Additionally, Fig. 6a shows glutathione
417
metabolism is significant but coverage level is not high, which may result from the
418
narrow detecting range provided by GC-MS method. This indicated that the metabolic
419
activation mediated by cytochromes P450 might be the triggering factors leading to
420
the development of EEA-induced liver injury.
421
Previous study showed that the DBT-induced hepatotoxicity had pronounced
422
impacts on the metabolism of amino acids (Zhao et al., 2017). Similarly, the results
423
from transcriptomics and metabonomics analyses (L-alanine, L-isoleucine, L-proline,
424
L-phenylalanine, and L-tyrosine) in our study supported this view that the metabolism
425
of amino acids played an important role in EEA-induced liver injury. Specifically, the
426
most notable changes were observed in taurine and hypotaurine metabolism. It should 20
427
be noticed that, compared with the control group, taurine was altered in the liver and
428
serum in response to EEA, while its content was increased in the serum and decreased
429
in the liver. Many studies have demonstrated that taurine was maintained in
430
abundance in the liver by both endogenous biosynthesis and exogenous transport,
431
though it decreased in liver diseases (Penttila et al., 1990; Matsuzaki et al., 1993;
432
Miyazaki and Matsuzaki, 2014). In addition, the most established role of taurine was
433
its conjugation with bile acids for excretion into bile (Danielsson, 1990). Therefore,
434
EEA-induced liver injuries might be strongly related to the reduction of taurine and
435
the disorder of BAs in the liver. The TCA cycle was an important hub of the
436
metabolism of carbohydrates, fats, and proteins, while L-malic acid, citric acid and
437
oxaloacetic acid were the dominant products of the TCA cycle (Vuoristo et al., 2016;
438
Zhang et al., 2016). As shown in Fig.5, L-malic acid and citric acid levels were
439
upgraded in EEA-treated groups, implying that there exists an imbalance with the
440
TCA cycle. Through further analysis, it was determined that TCA cycle functioned as
441
a bridge among the other disturbed metabolic pathways. Purine and pyrimidine
442
metabolisms might be the novel metabolic pathways for EEA-induced hepatotoxicity,
443
which was in agreement with our earlier work (Zhao et al., 2017). Taken in
444
combination, these results indicate that the EEA-induced liver injury might be
445
attributable in part ascribed to the imbalance of energy metabolism and metabolism of
446
amino acids.
447
Bile acid metabolism is referred to synthesis, transport and excretion (Halilbasic
448
et al., 2013). On one hand, the classical biosynthetic pathway (known as the neutral 21
449
BA biosynthetic pathway) is the primary pathway of BA biosynthesis. The present
450
RNA-Seq analysis showed that the expression of CYP7A1 (involved in BAs synthesis
451
from cholesterol) was significantly decreased, resulting in the reduction of BAs
452
biosynthesis (Russell, 2003). On the other hand, down-regulation was observed in the
453
expression of several important BAs transporters that joined in the hepatocellular
454
uptake: Na/taurocholate co-transporting polypeptide (NTCP/SLC10A1) and excretion
455
of BAs: bile salt export pump (BSEP), and the multidrug resistance protein 3 (MRP3)
456
(Soroka et al., 2014; Lam et al., 2010; Hirohashi et al., 2000). In general, we believed
457
that the metabolic balance of BAs was disturbed, thereby resulting in liver injury.
458
Thus, together with the protein analysis by western blotting analysis (Fig.8), these
459
results suggested that the imbalance of BAs metabolism might be responsible for
460
EEA-induced liver injury.
461
In summary, the integrative analysis of the gene expression and metabolites
462
levels changes following EEA exposure provide valuable information concerning the
463
hepatotoxic mechanism of action in different biological samples. In the process of 36
464
h administration of EEA (150 mg/kg), we found that the metabolic activation
465
mediated by cytochromes P450 might be the crucial steps in EEA-induced
466
hepatotoxicity, and the imbalance of energy metabolism, the metabolism of amino
467
acids and purine and pyrimidine metabolisms might also lead to the EEA-triggered
468
hepatic damage. This study suggested that the balance of BAs synthesis, uptake and
469
excretion was disturbed, the integrated effect on BAs metabolism resulting in liver
470
injury, and BAs including TCA and CA along with CDCA could be considered as 22
471
biomarkers of EEA-induced liver injury. The present findings will provide useful
472
information for further studies to examining the mechanism of EEA-induced
473
hepatotoxicity.
474
Acknowledgments
475
This work was supported by the National Natural Science Foundation of China
476
(No. 81773993) and the Project Funded by the Priority Academic Program
477
Development (PAPD) of Jiangsu Higher Education Institutions.
478
479
Conflicts of interest
480
The authors declare no conflicts of interest.
481
References
482
Afanador, N.L., Tran, T.N., Buydens, L.M., 2013. Use of the bootstrap and
483
permutation methods for a more robust variable importance in the projection
484
metric for partial least squares regression. Anal. Chim. Acta. 768, 49–56.
485
Basu, S., Duren, W., Evans, C.R., Burant, C.F., Michailidis, G., Karnovsky, A., 2017.
486
Sparse network modeling and metscape-based visualization methods for the
487
analysis of large-scale metabolomics data. Bioinformatics. 33, 1545–1553.
488
Bylesjö, M., Rantalainen, M., Cloarec, O., Nicholson, J. K., Holmes, E, Trygg, J.,
489
2010. OPLS discriminant analysis: combining the strengths of PLS-DA and
490
SIMCA classification. J. Chemometr. 20, 341–351.
491
Cui, Y.X., Paules, R.S., 2010. Use of transcriptomics in understanding mechanisms of 23
492
drug-induced toxicity. Pharmacogenomics 11, 573–585.
493
Chen, S., Zhang, M.Y., Bo, L., Li, S.Q., Hu, L.Y., Zhao, X.J., Sun, C.H., 2018.
494
Metabolomic analysis of the toxic effect of chronic exposure of cadmium on rat
495
urine. Environ. Sci. Pollut. R. 25, 3765–3774.
496
Chong, J., Soufan, O., Li, C., Caraus, I., Li, S., Bourque, G., Wishart, D.S., Xia, J.,
497
2018. MetaboAnalyst 4.0: towards more transparent and integrative metabolomics
498
analysis. Nucl. Acids. Res. 46, W486–W494.
499
Chan, E.C., Koh, P.K., Mal, M., Cheah, P.Y., Eu, K.W., Backshall A, Cavill, R.,
500
Nicholson, J.K., Keun, H.C., 2009. Metabolic Profiling of human colorectal cancer
501
using high-resolution magic angle spinning nuclear magnetic resonance (HR-MAS
502
NMR) spectroscopy and gas chromatography mass spectrometry (GC/MS). J.
503
Proteome. Res. 8, 352–361.
504
Dunn, W.B., Broadhurst, D., Begley, P., Zelena, E., Francis-McIntyre, S., Anderson,
505
N., Brown, M., Knowles, J.D., Halsall, A., Haselden, J.N., Nicholls, A.W., Wilson,
506
I.D., Kell, D.B., Goodacre, R., 2011. Procedures for large-scale metabolic
507
profiling of serum and serum using gas chromatography and liquid
508
chromatography coupled to mass spectrometry. Nat. Protoc. 6, 1060–1083.
509 510 511
Danielsson, H., 1963. Present status of research on catabolism and excretion of cholesterol. Adv. Lipid. Res. 1, 335–385. Fan, Y., Li, Y., Chen, Y., Zhao, Y.J., Liu, L.W., Li, J., Wang, S.L., Alolga, R.N., Yin, Y.,
512
Wang, X.M., Zhao, D.S., Shen, J.H., Meng, F.Q., Zhou, X., Xu, H., He, G.P., Lai,
513
M.D., Li, P., Zhu, W., Qi, L.W., 2016. Comprehensive metabolomic 24
514
characterization of coronary artery diseases. J. Am. Coll. Cardiol. 68, 1281–1293.
515
Fuchs, C.D., Paumgartner, G., Wahlström, A., Schwabl, P., Reiberger, T., Leditznig, N.,
516
Stojakovic,
517
BSEP/ABCB11−/− mice against cholestatic liver injury. J. hepatol. 66, 95–101.
518
Gao, H.Y., Kuroyanagi, M., Wu, L.J., Kawahara, N., Yasuno, T., Nakamura, Y., 2002.
519
Antitumor-promoting constituents from Dioscorea bulbifera L. in JB6 mouse
520
epidermal cells. Biol. Pharm. Bull. 25, 1241–1243.
T.,
Trauner,
M.,
2017.
Metabolic
preconditioning
protects
521
Huang, Z.F., Hua, B.C., Chen, X.F., Shi, D.H., Cheng, X.L., Wang, Y.H., Liao, H.J.,
522
2013. Analysis of liver injury in 78 cases caused by Rhizoma Dioscoreae
523
Bulbiferae and related preparation. Chin. J. Exp. Tradit. Med. Formulae 19,
524
295–297.
525
Hua, Y.L., Ji, P., Xue, Z.W., Wei, Y.M., 2015. Construction and analysis of correlation
526
networks based on gas chromatography-mass spectrometry metabonomics data for
527
lipopolysaccharide-induced inflammation and intervention with volatile oil from
528
Angelica sinensis in rats. Mol. BioSyst. 11, 3174–3187.
529
Hirohashi, T., Suzuki, H., Takikawa, H., Sugiyama, Y., 2000. ATP-dependent transport
530
of bile salts by rat multidrug resistance-associated protein 3 (Mrp3). J. Biol. Chem.
531
275, 2905–2910.
532 533
Halilbasic, E., Claudel, T., Trauner, M., 2013. Bile acid transporters and regulatory nuclear receptors in the liver and beyond. J. Hepatol. 58, 155–168.
534
Kawasaki, T., Komori, T., Setoguchi, S., 1968. Furanoid norditerpenes from
535
Dioscoreacae plants. 1. Diosbulins A, B, and C from Dioscorea bulbifera form a 25
536
spontanea. Chem. Pharm. Bull. 16, 2430–2435.
537
Katz, Y., Wang, E.T., Airoldi, E.M., Burge, C.B., 2010. Analysis and design of RNA
538
sequencing experiments for identifying isoform regulation. Nat. Methods 7,
539
1009–1015.
540
Liu, H., Tsim, K.W., Chou, G., Wang, J., Ji, L.L., Wang, Z.T., 2011. Phenolic
541
compounds from the rhizomes of Dioscorea bulbifera. Chem. Biodivers. 8,
542
2110–2116.
543 544
Liu, J.R., 2002. Two cases of toxic hepatitis caused by Dioscorea Bulbifera L. Adv. Drug React. 2, 129–130.
545
Lu, X.H., Wu, Q.H., 2014. Literature analysis of 33 cases of liver injury induced by
546
the compound preparation of Dioscorea bulbifera L. J. Sichuan Tradit. Chin. Med.
547
39, 162–164.
548
Lin, D.J., Guo, X.C., Gao, H.Y., Cheng, L., Cheng, M.S, Song, S., Peng, Y., Zheng, J.,
549
2015. In vitro and in vivo studies of the metabolic activation of 8-epidiosbulbin E
550
acetate. Chem. Res. Toxicol. 28, 1737–1746.
551
Lin, D.J., Li, W.W., Peng, Y., Jiang, C.F., Xu, Y.J., Gao, H.Y., Zheng, J., 2016. Role of
552
metabolic activation in 8-epidiosbulbin E acetate-induced liver injury: mechanism
553
of action of the hepatotoxic furanoid. Chem. Res. Toxicol. 29, 359–366.
554
Lin, D.J., Wang, K., Guo, X.C., Gao, H.Y., Peng, Y., Zheng, J., 2016. Lysine- and
555
cysteine-based
556
8-epidiosbulbin e acetate. Toxicol. Lett. 264, 20–28.
557
protein
adductions
derived
from
toxic
metabolites
of
Li, R.F., Yu, H.H., Xue, W., Yue, Y., Liu, S., Xing, R., Li, P.C., 2014. Jellyfish 26
558
venomics and venom gland transcriptomics analysis of Stomolophus meleagris to
559
reveal the toxins associated with sting. J. Proteomics. 106, 17–29.
560
Lu, X.Y., Hu, B., Shao, L., Tian, Y., Jin, T.T., Jin, Y.H., Ji, S., Fan, X.H., 2013.
561
Integrated analysis of transcriptomics and metabonomics profiles in aflatoxin
562
B1-induced hepatotoxicity in rat. Food Chem. Toxicol. 55, 444–455.
563 564
Livak, K.J., Schmittgen, T.D., 2001. Analysis of relative gene expression data using real-time quantitative PCR and the 2−∆∆CT method. Methods 25, 402–408.
565
Lam, P., Soroka, C.J., Boyer, J.L., 2010. The bile salt export pump: clinical and
566
experimental aspects of genetic and acquired cholestatic liver disease. Semi. Liver
567
Dis. 30, 125–133.
568
Murray, R.D.H., Jorge, Z.D., Khan, N.H., Shahjahan, M., Quaisuddin, M., 1984.
569
Diosbulbin D and 8-epidiosbulbin E acetate, norclerodane diterpenoids from
570
Dioscorea bulbifera tubers. Phytochemistry 23, 623–625.
571
Matthews, D.R., Hosker, J.P., Rudenski, A.S., Naylor, B.A., Treacher, D.F., Turner,
572
R.C., 1985. Homeostasis model assessment: insulin resistance and beta-cell
573
function from fasting serum glucose and insulin concentrations in man.
574
Diabetologia 28, 412–419.
575
McEuen, K., Borlak, J., Tong, W.D., Chen, M.J., 2017. Associations of drug
576
lipophilicity and extent of metabolism with drug-induced liver injury. Int. J. Mol.
577
Sci. 18, 1335.
578
Matsuzaki, Y., Tanaka, N., Osuga, T., 1993. Is taurine effective for treatment of
579
painful muscle cramps in liver cirrhosis? Am. J. Gastroenterol. 88, 1466–1467. 27
580 581 582 583
Miyazaki, T., Matsuzaki, Y., 2014. Taurine and liver diseases: a focus on the heterogeneous protective properties of taurine. Amino. Acids 46, 101–110. Penttila, K.E., 1990. Role of cysteine and taurine in regulating glutathione synthesis by periportal and perivenous hepatocytes. Biochem. J. 269, 659–664.
584
Qu, X.Y., Tao, L.N., Zhang, S.X., Sun, J.M., Niu, J.Q., Ding Y.H., Song Y.Q., 2017.
585
The role of Ntcp, Oatp2, Bsep and Mrp2 in liver injury induced by Dioscorea
586
bulbifera L. and diosbulbin B in mice. Environ. Toxicol. Pharmacol. 51, 16–22.
587
Rao, Y.Z., Wen, T.L., Zhao, H.M., 2010. Study on anti-inflammatory effect of
588
methanol extracts from Dioscorea bulbifera L. Anhui Agri. Sci. Bull. 16, 64–65.
589
Renaud, H.J., Cui, J., Khan, M., Klaassen, C.D., 2011. Tissue distribution and
590
gender-divergent expression of 78 cytochrome P450 mRNAs in mice. Toxicol. Sci.
591
124, 261–277.
592 593 594 595
Ramadori, P., Weiskirchen, R., Trebicka, J., Streetz, K., 2015. Mouse models of metabolic liver injury. Lab. Anim. 49, 47–58. Russell, D.W., 2003. The enzymes, regulation, and genetics of bile acid synthesis. Annu. Rev. Biochem. 72, 137–174.
596
Shi, W., Zhang, C., Zhao, D.S., Wang, L.L., Li P., Li, H.J., 2018. Discovery of
597
hepatotoxic equivalent combinatorial markers from Dioscorea bulbifera tuber by
598
fingerprint-toxicity relationship modeling. Sci. Rep. 8, 462.
599
Suo, Y.J., Gao, S.G., Baranzoni, G.M., Xie, Y.P., Liu, Y.H., 2018. Comparative
600
transcriptome rna-seq analysis of listeria monocytogenes, with sodium lactate
601
adaptation. Food Control 91, 193–201. 28
602
Soroka, C.J., Boyer, J.L., 2014. Biosynthesis and trafficking of the bile salt export
603
pump, BSEP: therapeutic implications of BSEP mutations. Mol. Aspects Med. 37,
604
3–14.
605 606
Tang, Y.X., 1995. The research of Dioscoreae bulbifera L. in clinical application. Chin J Chin. Mater. Med. 20, 435–438.
607
Trapnell, C., Williams, B.A., Pertea, G., Mortazavi, A., Kwan, G., van, Baren M.J.,
608
Salzberg, S.L., Wold, B.J., Pachter, L., 2010. Transcript assembly and
609
quantification by RNA-Seq reveals unannotated transcripts and isoform switching
610
during cell differentiation. Nat. Biotechnol. 28, 511–515.
611
Van Swelm, R.P., Kramers, C., Masereeuw, R., Russel, F.G., 2014. Application of
612
urine proteomics for biomarker discovery in drug-induced liver injury. Crit. Rev.
613
Toxicol. 44, 823–841.
614
Vuoristo, K.S., Mars, A.E., Sanders, J.P.M., Eggink, G., Weusthuis, R.A., 2016.
615
Metabolic engineering of TCA cycle for production of chemicals. Trends
616
Biotechnol. 34, 191–197.
617
Wang, J.M., Ji, L.L., Branford-White, C.J., Wang, Z.Y., Shen, K.K, Liu, H., Wang,
618
Z.T., 2012. Antitumor activity of Dioscorea bulbifera L. Rhizome in vivo.
619
Fitoterapia 83, 388–394.
620 621
Wang, J.M., Ji L.L., Liu, H., Wang, Z.T., 2010. Study of the hepatotoxicity induced by Dioscorea bulbifera L. rhizome in mice. BioSci. Trends 4, 79–85.
622
Wang, J.M., Liang, Q.N., Ji, L.L, Liu, H., Wang, C.H., Wang, Z.T., 2011.
623
Gender-related difference in liver injury induced by Dioscorea bulbifera L. 29
624
rhizome in mice. Hum. Exp. Toxicol. 30, 1333–1341.
625
Whitfield, J.B., Martin, N.G., 1985. Individual differences in serum ALT, AST and
626
GGT: contributions of genetic and environmental factors, including alcohol
627
consumption. Enzyme 33, 61–69.
628
Xiong, Y.H., Xu, Y.U., Yang, L., Wang, Z.T., 2017. The metabolic profile of serum
629
fatty acids in rats with Rhizoma Dioscoreae Bulbiferae-induced liver injury. Acta.
630
Pharm. Sin. 52, 753–759.
631
Yang, H., Li, Y.X., Cui, X.Q., Yang, C., Li, L.Y., Liu, J.L., Mu, L.C., Yuan, J.C.,
632
Zhang, B., 2006. Clinical use and adverse drug reaction of compound prescription
633
of Dioscorea bulbifera L. in clinical trial. Clin. Misdiagn. Misther. 19, 85–87.
634
Zhao, D.S., Jiang, L.L., Fan, Y.X., Wang, L.L., Li, Z.Q., Shi, W, Li, P, Li, H.J., 2017.
635
Investigation of Dioscorea bulbifera rhizome-induced hepatotoxicity in rats by a
636
multisample integrated metabolomics approach. Chem. Res. Toxicol. 30,
637
1865–1873.
638
Zhao, D.S., Jiang, L.L., Fan, Y.X., Dong, L.C., Ma, J., Dong, X., Xu, X.J., Li, P., Li,
639
H.J., 2017. Identification of urine tauro-β-muricholic acid as a promising
640
biomarker in Polygoni Multiflori Radix-induced hepatotoxicity by targeted
641
metabolomics of bile acids. Food Chem. Toxicol. 108, 532–542.
642
Zhang, C.E., Niu, M., Li, Q., Zhao, Y.L., Ma, Z.J., Xiong, Y., Dong, X.P., Li, R.Y.,
643
Feng, W.W., Dong, Q., Ma, X., Zhu, Y., Zou, Z.S., Cao, J.L., Wang, J.B., Xiao,
644
X.H., 2016. Urine metabolomics study on the liver injury in rats induced by raw
645
and processed Polygonum multiflorum integrated with pattern recognition and 30
646
pathways analysis. J. Ethnopharmacol. 194, 299–306.
647
31
648
Figure captions
649
Fig. 1 Summary figure representing the experimental design, methods and tools
650
applied to investigate EEA-induced hepatotoxicity in mice using the integrated
651
transcriptomic and metabolomics method. EEA: 8-epidiosbulbin E acetate.
652 653
Fig. 2 Typical liver histopathology (A: control group; B: EEA-treated group) and
654
biochemical parameters (C-G, ALT/AST/TBIL/DBIL/ALP activities) in mice 36 h
655
following i.g. administration of EEA at 0, 30, or 150 mg/kg. Results of quantitative
656
analysis values are expressed as mean ± SD (n = 6). Significant differences from the
657
value of control group with the administration of EEA were noted (*p < 0.05, **p <
658
0.01).
659 660
Fig. 3 GC-TOF MS profiles of serum samples in EEA (150 mg/kg) and control
661
groups. Principal component analysis (PCA) model (A) and the pattern recognition of
662
orthogonal partial least-squares-discriminate analysis (OPLS-DA) model (B/C). The
663
green circle represents control group, the blue circle represents EEA-treated group,
664
the red circle represents quality control(QC) group, respectively.
665 666
Fig. 4 GC-TOF MS profiles of liver tissue samples in EEA (150 mg/kg) and control
667
groups. PCA model (A) and the pattern recognition of OPLS-DA model (B/C).
668 669
Fig. 5 Heat map visualization of differentially abundant metabolites in serum (A) and 32
670
liver tissue (B) samples of EEA (150 mg/kg)-administered and control groups, Colors
671
from highest (red) to lowest (blue) represent metabolite expression values in different
672
groups.
673 674
Fig. 6 Analysis of metabolic disorders in serum (A) and liver tissue (B) in EEA (150
675
mg/kg)-treated groups and control groups using MetaboAnalyst 4.0. Bubble size and
676
color show the significance of path arrangement. The lighter and smaller bubbles
677
represent least affected pathways, whereas the larger and darker bubbles represent the
678
more markedly affected pathways.
679 680
Fig. 7 A summary of the result from the integrated pathway analysis module of
681
transcriptome and metabolome in EEA (150 mg/kg)-treated mice. The stacked bars
682
show the accumulated contributions from enrichment and topology analysis.
683 684
Fig. 8 Western verification results of differentially expressed genes (FN1, UGT1A1,
685
CYP1B1, CYP1A1, CYP2E1, CYP8B1, CYP7A1, BAAT, NTCP, BSEP and MRP3)
686
in the livers of mice treated with EEA at 30, or 150 mg/kg. β-actin level was used as
687
the internal reference. Vehicle: control group, EEA-30: EEA (30 mg/kg)-treated group,
688
EEA-150: EEA (150 mg/kg)-treated group.
689 690
Fig. 9 Alterations of the BAs concentrations in response to EEA (150 mg/kg)
691
consumption. The data are expressed as the mean ± SD (n = 6). Significant 33
692
differences from the value of control group with the administration of EEA were
693
indicated (*p < 0.05, **p < 0.01). Vehicle: control group, EEA-150: EEA (150
694
mg/kg)-treated group.
695
34
696
Table 1 The quantitative PCR primers of candidate genes. Number
Gene name
1
CYP7A1
2
BAAT
3
FH1
4
ABCC2
5
SlCO1A4
6
CYP2E1
7
ADA
8
IDH2
9
UGT1A1
10
CYP3A25
Primer sequence S: TTCATCACAAACTCCCTGTCATAC A: TTCCATCACTTGGGTCTATGCT S: ATGAATAGCCCCTACCAAATCC A: TCCACCAGCACCTCCAAACA S: AGATTGGAGGTGCTACGGAACG A: TCCAGTCTGCCAAACCACCA S: CATTGGCTTCGTGAAAGACCCT A: AATCGTGTACTGCCTCCTAGCC S: GGGTTGCCTGCTGCTCTAAGA A: TTCCGTTCTCCATCATTCTGCA S: AGGCTGTCAAGGAGGTGCTAC A: GCACAGCCAATCAGAAAGGTAG S: CAGACACCCGCATTCAACAA A: TGTCCATGCCGATAATGTTGC S: ACAGTCACCCGCCATTACCG A: TCCAGCGTCTGTGCAAACCT S: ACGCTGGGAGGCTGTTAGT A: CCGTCCAAGTTCCACCAAAG S: GGAGGCCTGAACTGCTAAAG A: GTAGTTGAAAATGGTGCCAAGTAAC
697
35
Highlights: An integrated transcriptomic/metabolomics method for screening EEA hepatotoxicity Purine and pyrimidine metabolisms might be the novel metabolic pathways for EEA-induced hepatotoxicity Imbalance of bile acid metabolism might be responsible for EEA-induced hepatotoxicity TCA and CA along with CDCA could be considered as biomarkers
Declaration of interests ☐ The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper. ☐The authors declare the following financial interests/personal relationships which may be considered as potential competing interests:
The authors declare that there is no conflict of interests regarding the publication of this article.